Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.459
Filter
1.
Bioact Mater ; 43: 195-224, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39386221

ABSTRACT

The epithelium is one of the important tissues in the body as it plays a crucial barrier role serving as a gateway into and out of the body. Most organs in the body contain an epithelial tissue component, where the tightly connected, organ-specific epithelial cells organize into cysts, invaginations, or tubules, thereby performing distinct to endocrine or exocrine secretory functions. Despite the significance of epithelium, engineering functional epithelium in vitro has remained a challenge due to it is special architecture, heterotypic composition of epithelial tissues, and most importantly, difficulty in attaining the apico-basal and planar polarity of epithelial cells. Bioprinting has brought a paradigm shift in fabricating such apico-basal polarized tissues. In this review, we provide an overview of epithelial tissues and provide insights on recapitulating their cellular arrangement and polarization to achieve epithelial function. We describe the different bioprinting techniques that have been successful in engineering polarized epithelium, which can serve as in vitro models for understanding homeostasis and studying diseased conditions. We also discuss the different attempts that have been investigated to study these 3D bioprinted engineered epithelium for preclinical use. Finally, we highlight the challenges and the opportunities that need to be addressed for translation of 3D bioprinted epithelial tissues towards paving way for personalized healthcare in the future.

2.
Regen Biomater ; 11: rbae094, 2024.
Article in English | MEDLINE | ID: mdl-39350955

ABSTRACT

To address the uneven nutrient distribution within three-dimensional (3D) tissue models and organoids currently used in medical research, this study introduces a microvascular network based on the Hilbert curve. Our aim was to develop innovative solutions for enhancing nutrient supply in thick tissue models in vitro. By using 3D bioprinting, we engineered microvascular networks of varying Hilbert orders and validated their efficacy in enhancing nutrient uniformity through numerical simulations and experiments. These networks facilitated broader and more uniform nutrient distribution throughout the thick tissue models, particularly the 2° Hilbert microvascular structure, which occupies less space and significantly reduces regions of cellular death. Furthermore, we explored the potential of assembling larger tissue constructs using the 2° Hilbert microvascular network, showcasing its applicability in constructing large-scale biological models. The findings suggest that the 2° Hilbert microvascular structure is particularly effective in ensuring adequate nutrient delivery, thus enhancing the viability and functionality of large-volume tissue models. These innovations hold significant promise for advancing the fields of tissue engineering and regenerative medicine by improving nutrient delivery to in vitro thick tissue block models. This provides a robust foundation for future in vitro research and clinical applications, potentially leading to more effective treatments and interventions in the medical field. The development of these microvascular networks represents a crucial step forward in overcoming the limitations of current 3D tissue models and organoids, paving the way for more sophisticated and reliable biomedical research tools.

4.
J Mol Med (Berl) ; 2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39358606

ABSTRACT

Wound healing, an intricate biological process, comprises orderly phases of simple biological processed including hemostasis, inflammation, angiogenesis, cell proliferation, and ECM remodeling. The regulation of the shift in these phases can be influenced by systemic or environmental conditions. Any untimely transitions between these phases can lead to chronic wounds and scarring, imposing a significant socio-economic burden on patients. Current treatment modalities are largely supportive in nature and primarily involve the prevention of infection and controlling inflammation. This often results in delayed healing and wound complications. Recent strides in regenerative medicine and tissue engineering offer innovative and patient-specific solutions. Mesenchymal stem cells (MSCs) and their secretome have gained specific prominence in this regard. Additionally, technologies like tissue nano-transfection enable in situ gene editing, a need-specific approach without the requirement of complex laboratory procedures. Innovating approaches like 3D bioprinting and ECM bioscaffolds also hold the potential to address wounds at the molecular and cellular levels. These regenerative approaches target common healing obstacles, such as hyper-inflammation thereby promoting self-recovery through crucial signaling pathway stimulation. The rationale of this review is to examine the benefits and limitations of both current and emerging technologies in wound care and to offer insights into potential advancements in the field. The shift towards such patient-centric therapies reflects a paradigmatic change in wound care strategies.

5.
Adv Mater ; : e2412127, 2024 Oct 10.
Article in English | MEDLINE | ID: mdl-39385640

ABSTRACT

Embedded 3D bioprinting techniques have emerged as a powerful method to fabricate 3D engineered constructs using low strength bioinks; however, there are challenges in simultaneously satisfying the requirements of high-cell-activity, high-cell-proportion, and low-viscosity bioinks. In particular, the printing capacity of embedded 3D bioprinting is limited as two main challenges: spreading and diffusion, especially for liquid, high-cell-activity bioinks that can facilitate high-cell-proportion. Here, a liquid-in-liquid 3D bioprinting (LL3DBP) strategy is developed, which used a liquid granular bath to prevent the spreading of liquid bioinks during 3D printing, and electrostatic interaction between the liquid bioinks and liquid granular baths is found to effectively prevent the diffusion of liquid bioinks. As an example, the printing of positively charged 5% w/v gelatin methacryloyl (GelMA) in a liquid granular bath prepared with negatively charged κ-carrageenan is proved to be achievable. By LL3DBP, printing capacity is greatly advanced and bioinks with over 90% v/v cell can be printed, and printed structures with high-cell-proportion exhibit excellent bioactivity.

6.
Adv Funct Mater ; 34(28)2024 Jul 10.
Article in English | MEDLINE | ID: mdl-39380942

ABSTRACT

3-D bioprinting is a promising technology to fabricate custom geometries for tissue engineering. However, most bioprintable hydrogels are weak and fragile, difficult to handle and cannot mimetic the mechanical behaviors of the native soft elastic tissues. We have developed a visible light crosslinked, single-network, elastic and biocompatible hydrogel system based on an acrylated triblock copolymer of poly(ethylene glycol) PEG and polycaprolactone (PCL) (PEG-PCL-DA). To enable its application in bioprinting of soft tissues, we have modified the hydrogel system on its printability and biodegradability. Furthermore, we hypothesize that this elastic material can better transmit pulsatile forces to cells, leading to enhanced cellular response under mechanical stimulation. This central hypothesis was tested using vascular conduits with smooth muscle cells (SMCs) cultured under pulsatile forces in a custom-made bioreactor. The results showed that vascular conduits made of PEG-PCL-DA hydrogel faithfully recapitulate the rapid stretch and recoil under the pulsatile pressure from 1 to 3 Hz frequency, which induced a contractile SMC phenotype, consistently upregulated the core contractile transcription factors. In summary, our work demonstrates the potential of elastic hydrogel for 3D bioprinting of soft tissues by fine tuning the printability, biodegradability, while possess robust elastic property suitable for manual handling and biomechanical stimulation.

7.
Biofabrication ; 2024 Oct 04.
Article in English | MEDLINE | ID: mdl-39366424

ABSTRACT

Intervertebral disc (IVD) function is achieved through integration of its two component regions: the nucleus pulposus (NP) and the annulus fibrosus (AF). The NP is soft (0.3-5 kPa), gelatinous and populated by spherical NP cells in a polysaccharide-rich extracellular matrix (ECM). The AF is much stiffer (~100 kPa) and contains layers of elongated AF cells in an aligned, fibrous ECM. Degeneration of the disc is a common problem with age being a major risk factor. Progression of IVD degeneration leads to chronic pain and can result in permanent disability. The development of therapeutic solutions for IVD degeneration is impaired by a lack of in vitro models of the disc that are capable of replicating the fundamental structure and biology of the tissue. This study aims to investigate if a newly developed suspended hydrogel bioprinting system (termed SLAM) could be employed to fabricate IVD analogues with integrated structural and compositional features similar to native tissue. Bioprinted IVD analogues were fabricated to recapitulate structural, morphological and biological components present in the native tissue. The constructs replicated key structural components of native tissue with the presence of a central, polysaccharide-rich NP surrounded by organised, aligned collagen fibres in the AF. Cell tracking, actin and matrix staining demonstrated that embedded NP and AF cells exhibited morphologies and phenotypes analogous to what is observed in vivo with elongated, aligned AF cells and spherical NP cells that deposited HA into the surrounding environment. Critically, it was also observed that the NP and AF regions contained a defined cellular and material interface and segregated regions of the two cell types, thus mimicking the highly regulated structure of the IVD.

8.
Sci Rep ; 14(1): 23240, 2024 10 05.
Article in English | MEDLINE | ID: mdl-39369014

ABSTRACT

Gelatin methacryloyl (GelMA), typically derived from mammalian sources, has recently emerged as an ideal bio-ink for three-dimensional (3D) bioprinting. Herein, we developed a fish skin-based GelMA bio-ink for the fabrication of a 3D GelMA skin substitute with a 3D bioprinter. Several concentrations of methacrylic acid anhydride were used to fabricate GelMA, in which their physical-mechanical properties were assessed. This fish skin-based GelMA bio-ink was loaded with human adipose tissue-derived mesenchymal stromal cells (ASCs) and human platelet lysate (HPL) and then printed to obtain 3D ASCs + HPL-loaded GelMA scaffolds. Cell viability test and a preliminary investigation of its effectiveness in promoting wound closure were evaluated in a critical-sized full thickness skin defect in a rat model. The cell viability results showed that the number of ASCs increased significantly within the 3D GelMA hydrogel scaffold, indicating its biocompatibility property. In vivo results demonstrated that ASCs + HPL-loaded GelMA scaffolds could delay wound contraction, markedly enhanced collagen deposition, and promoted the formation of new blood vessels, especially at the wound edge, compared to the untreated group. Therefore, this newly fish skin-based GelMA bio-ink developed in this study has the potential to be utilized for the printing of 3D GelMA skin substitutes.


Subject(s)
Bioprinting , Gelatin , Mesenchymal Stem Cells , Printing, Three-Dimensional , Skin, Artificial , Tissue Scaffolds , Gelatin/chemistry , Animals , Bioprinting/methods , Humans , Rats , Tissue Scaffolds/chemistry , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Fishes , Methacrylates/chemistry , Skin/metabolism , Skin/drug effects , Ink , Wound Healing/drug effects , Tissue Engineering/methods , Cell Survival/drug effects , Hydrogels/chemistry , Adipose Tissue/cytology , Adipose Tissue/metabolism , Biocompatible Materials/chemistry
9.
Article in English | MEDLINE | ID: mdl-39388391

ABSTRACT

The establishment of organotypic preclinical models that accurately resemble the native tumor microenvironment at an anatomic human scale is highly desirable to level up in vitro platforms potential for screening candidate therapies. The bioengineering of anatomic-scaled three-dimensional (3D) models that emulate native tumor scale while recapitulating their cellular and matrix components remains, however, to be fully realized. In this focus, herein, we leveraged embedded 3D bioprinting for biofabricating pancreatic ductal adenocarcinoma (PDAC) in vitro models combining gelatin-methacryloyl and hyaluronic acid methacrylate extracellular matrix (ECM)-mimetic biomaterials with human pancreatic cancer cells and cancer-associated fibroblasts to generate in vitro models capable of emulating native tumor size (∼6 mm) and stromal elements. By using a viscoelastic continuous polymeric supporting bath, tumor-scale 3D models were rapidly generated (∼50 constructs/h) and easily recovered following in-bath visible light photocrosslinking. As a proof-of-concept, tissue-scale constructs displaying physiomimetic designs were biofabricated. These models also encompass the incorporation of a stromal compartment to better emulate the cellular components of the PDAC native tumor microenvironment (TME) and its stratified spatial organization. Cell-laden tumor-size constructs remained viable for up to 14 days and were responsive to Gemcitabine in a dose-dependent mode. Cancer-stroma models also exhibited increased drug resistance compared to their monotypic counterparts, highlighting the key role of stromal cells in chemotherapeutic resistance. Overall, we report for the first time the freeform biofabrication of PDAC models exhibiting anatomic scale, different structural complexities, and engineered cancer-stromal compartments, being highly valuable for preclinical screening of therapeutics.

10.
EXCLI J ; 23: 1170-1182, 2024.
Article in English | MEDLINE | ID: mdl-39391058

ABSTRACT

Cartilage tissue, characterized by its limited regenerative capacity, presents significant challenges in clinical therapy. Recent advancements in cartilage regeneration have focused on integrating stem cell therapies, tissue engineering strategies, and advanced modeling techniques to overcome existing limitations. Stem cells, particularly Mesenchymal Stem Cells (MSCs) and induced pluripotent stem cells (iPSCs), hold promise for cartilage repair due to their ability to differentiate into chondrocytes, the key cells responsible for cartilage formation. Tissue engineering approaches, including 3D models, organ-on-a-chip systems, and organoids, offer innovative methods to mimic natural tissue microenvironments and evaluate potential treatments. MSC-based techniques, such as cell sheet tissue engineering, address challenges associated with traditional therapies, including cell availability and culture difficulties. Furthermore, advancements in 3D bioprinting enable the fabrication of complex tissue structures, while organ-on-a-chip systems provide microfluidic platforms for disease modeling and physiological mimicry. Organoids serve as simplified models of organs, capturing some complexity and enabling the monitoring of pathophysiological aspects of cartilage diseases. This comprehensive review underscores the transformative potential of integrating stem cell therapies, tissue engineering strategies, and advanced modeling techniques to improve cartilage regeneration and pave the way for more effective clinical treatments.

11.
Sci Technol Adv Mater ; 25(1): 2404382, 2024.
Article in English | MEDLINE | ID: mdl-39328923

ABSTRACT

Current in vitro and in vivo tests applied to assess the safety of medical devices retain several limitations, such as an incomplete ability to faithfully recapitulate human features, and to predict the response of human tissues together with non-trivial ethical aspects. We here challenged a new hybrid biofabrication technique that combines bioprinting and Fast Diffusion-induced Gelation strategy to generate a vessel-like structure with the attempt to spatially organize fibroblasts, smooth-muscle cells, and endothelial cells. The introduction of Fast Diffusion-induced Gelation minimizes the endothelial cell mortality during biofabrication and produce a thin endothelial layer with tunable thickness. Cell viability, Von Willebrand factor, and CD31 expression were evaluated on biofabricated tissues, showing how bioprinting and Fast Diffusion-induced Gelation can replicate human vessels architecture and complexity. We then applied biofabricated tissue to study the cytotoxicity of a carbothane catheter under static condition, and to better recapitulate the effect of blood flow, a novel bioreactor named CuBiBox (Customized Biological Box) was developed and introduced in a dynamic modality. Collectively, we propose a novel bioprinted platform for human in vitro biocompatibility testing, predicting the impact of medical devices and their materials on vascular systems, reducing animal experimentation and, ultimately, accelerating time to market.


Our study provides an innovative convergence of 3D biofabrication technologies to realize multi-cellularized vessel-like models, as a new tool for in vitro biocompatibility testing of medical devices, minimizing animal experimentation.

12.
J Artif Organs ; 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39327399

ABSTRACT

Improvements in the roll porous scaffold (RPS) 3D bioproduction technology will increase print density of 10-15 µm cells by ~ 20% up to ~ 1.5 × 108 cells/mL and purity of organoid formation by > 17%. The use of 360 and 1200 dpi inkjet printheads immediately enables biomanufacturing with 10-30 µm cells in a single organoid with performance > 1.8 L/h for 15 µm layer thickness. The spongy bioresorbable ribbon for RPS technology is designed to solve the problems of precise placement, leakage and increasing in the number of instantly useable cell types and superior to all currently dominant 3D bioprinting methods in speed, volume, and print density without the use of expensive equipment and components. The potential of RPS for parallel testing of new substances studied was not on animals, but using generated 3D biomodels "organ on a chip". Solid organoids are more suitable for personalized medicine with simultaneous checking of several treatment methods and drugs, targeted therapy for a specific patient in vitro using the 3D composition of his personal cells, and selection of the most effective ones with the least toxicity. Overcoming the shortage of organs for implantation and personal hormone replacement therapy for everyone was achieved using printed endocrine glands based on their DNA.

13.
Biomolecules ; 14(9)2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39334833

ABSTRACT

Skin aging is influenced by intrinsic and extrinsic factors that progressively impair skin functionality over time. Investigating the skin aging process requires thorough research using innovative technologies. This review explores the use of in vitro human 3D culture models, serving as valuable alternatives to animal ones, in skin aging research. The aim is to highlight the benefits and necessity of improving the methodology in analyzing the molecular mechanisms underlying human skin aging. Traditional 2D models, including monolayers of keratinocytes, fibroblasts, or melanocytes, even if providing cost-effective and straightforward methods to study critical processes such as extracellular matrix degradation, pigmentation, and the effects of secretome on skin cells, fail to replicate the complex tissue architecture with its intricated interactions. Advanced 3D models (organoid cultures, "skin-on-chip" technologies, reconstructed human skin, and 3D bioprinting) considerably enhance the physiological relevance, enabling a more accurate representation of skin aging and its peculiar features. By reporting the advantages and limitations of 3D models, this review highlights the importance of using advanced in vitro systems to develop practical anti-aging preventive and reparative approaches and improve human translational research in this field. Further exploration of these technologies will provide new opportunities for previously unexplored knowledge on skin aging.


Subject(s)
Skin Aging , Humans , Skin Aging/physiology , Skin/metabolism , Melanocytes/metabolism , Melanocytes/cytology , Keratinocytes/cytology , Keratinocytes/metabolism , Fibroblasts/metabolism , Fibroblasts/cytology , Models, Biological , Printing, Three-Dimensional , Bioprinting/methods , Cell Culture Techniques, Three Dimensional/methods
14.
J Tissue Eng ; 15: 20417314241282476, 2024.
Article in English | MEDLINE | ID: mdl-39345255

ABSTRACT

Three-dimensional (3D) bioprinting has emerged as a promising strategy for fabricating complex tissue analogs with intricate architectures, such as vascular networks. Achieving this necessitates bioink formulations that possess highly printable properties and provide a cell-friendly microenvironment mimicking the native extracellular matrix. Rapid advancements in printing techniques continue to expand the capabilities of researchers, enabling them to overcome existing biological barriers. This review offers a comprehensive examination of ultraviolet-based 3D bioprinting, renowned for its exceptional precision compared to other techniques, and explores its applications in inducing angiogenesis across diverse tissue models related to hypoxia. The high-precision and rapid photocuring capabilities of 3D bioprinting are essential for accurately replicating the intricate complexity of vascular networks and extending the diffusion limits for nutrients and gases. Addressing the lack of vascular structure is crucial in hypoxia-related diseases, as it can significantly improve oxygen delivery and overall tissue health. Consequently, high-resolution 3D bioprinting facilitates the creation of vascular structures within three-dimensional engineered tissues, offering a potential solution for addressing hypoxia-related diseases. Emphasis is placed on fundamental components essential for successful 3D bioprinting, including cell types, bioink compositions, and growth factors highlighted in recent studies. The insights provided in this review underscore the promising prospects of leveraging 3D printing technologies for addressing hypoxia-related diseases through the stimulation of angiogenesis, complementing the therapeutic efficacy of cell therapy.

15.
Article in English | MEDLINE | ID: mdl-39309029

ABSTRACT

The demand for meat and seafood products has been globally increasing for decades. To address the environmental, social, and economic impacts of this trend, there has been a surge in the development of three-dimensional (3D) food bioprinting technologies for lab-grown muscle food products and their analogues. This innovative approach is a sustainable solution to mitigate the environmental risks associated with climate change caused by the negative impacts of indiscriminative livestock production and industrial aquaculture. This review article explores the adoption of 3D bioprinting modalities to manufacture lab-grown muscle food products and their associated technologies, cells, and bioink formulations. Additionally, various processing techniques, governing the characteristics of bioprinted food products, nutritional compositions, and safety aspects as well as its relevant ethical and social considerations, were discussed. Although promising, further research and development is needed to meet standards and translate into several industrial areas, such as the food and renewable energy industries. In specific, optimization of animal cell culture conditions, development of serum-free media, and bioreactor design are essential to eliminate the risk factors but achieve the unique nutritional requirements and consumer acceptance. In short, the advancement of 3D bioprinting technologies holds great potential for transforming the food industry, but achieving widespread adoption will require continued innovation, rigorous research, and adherence to ethical standards to ensure safety, nutritional quality, and consumer acceptance.

16.
Polymers (Basel) ; 16(18)2024 Sep 22.
Article in English | MEDLINE | ID: mdl-39339133

ABSTRACT

Biomaterials have been the subject of extensive research, and their applications in medicine and pharmacy are expanding rapidly. Collagen and its derivatives stand out as valuable biomaterials due to their high biocompatibility, biodegradability, and lack of toxicity and immunogenicity. This review comprehensively examines collagen from various sources, its extraction and processing methods, and its structural and functional properties. Preserving the native state of collagen is crucial for maintaining its beneficial characteristics. The challenges associated with chemically modifying collagen to tailor its properties for specific clinical needs are also addressed. The review discusses various collagen-based biomaterials, including solutions, hydrogels, powders, sponges, scaffolds, and thin films. These materials have broad applications in regenerative medicine, tissue engineering, drug delivery, and wound healing. Additionally, the review highlights current research trends related to collagen and its derivatives. These trends may significantly influence future developments, such as using collagen-based bioinks for 3D bioprinting or exploring new collagen nanoparticle preparation methods and drug delivery systems.

17.
Sheng Wu Gong Cheng Xue Bao ; 40(9): 2934-2947, 2024 Sep 25.
Article in Chinese | MEDLINE | ID: mdl-39319716

ABSTRACT

Cardiovascular diseases are major diseases, and there is lack of artificial blood vessels with small diameters which can be applied in coronary artery bypass surgery. The conventional vascular scaffold preparation techniques in tissue engineering have shortcomings in regulating the diameter, geometric shape, and interconnectivity of the scaffold. 3D bioprinting can simulate the natural structure of the vascular tissue, accurately print live cells and biomaterials, and regulate the microstructure and porosity of scaffolds on the nanoscale, providing new ideas for vascular tissue engineering. This article systematically evaluates the classification of 3D bioprinting technologies and reviews the latest research progress of 3D bioprinting in vascular tissue engineering. It summarizes the advantages of 3D bioprinting and points out the problems that need to be solved, such as the immune rejection of blood vessel materials, providing reference for the further research.


Subject(s)
Bioprinting , Printing, Three-Dimensional , Tissue Engineering , Tissue Scaffolds , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Humans , Blood Vessels , Biocompatible Materials , Blood Vessel Prosthesis
18.
Adv Funct Mater ; 34(7)2024 Feb.
Article in English | MEDLINE | ID: mdl-39257639

ABSTRACT

The availability of grafts to replace small-diameter arteries remains an unmet clinical need. Here, the validated methodology is reported for a novel hybrid tissue-engineered vascular graft that aims to match the natural structure of small-size arteries. The blood vessel mimic (BVM) comprises an internal conduit of co-electrospun gelatin and polycaprolactone (PCL) nanofibers (corresponding to the tunica intima of an artery), reinforced by an additional layer of PCL aligned fibers (the internal elastic membrane). Endothelial cells are deposited onto the luminal surface using a rotative bioreactor. A bioprinting system extrudes two concentric cell-laden hydrogel layers containing respectively vascular smooth muscle cells and pericytes to create the tunica media and adventitia. The semi-automated cellularization process reduces the production and maturation time to 6 days. After the evaluation of mechanical properties, cellular viability, hemocompatibility, and suturability, the BVM is successfully implanted in the left pulmonary artery of swine. Here, the BVM showed good hemostatic properties, capability to withstand blood pressure, and patency at 5 weeks post-implantation. These promising data open a new avenue to developing an artery-like product for reconstructing small-diameter blood vessels.

19.
Adv Healthc Mater ; : e2402075, 2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39313990

ABSTRACT

The muscle-tendon junction (MTJ) plays a pivotal role in efficiently converting the muscular contraction into a controlled skeletal movement through the tendon. Given its complex biomechanical intricacy, the biofabrication of such tissue interface represents a significant challenge in the field of musculoskeletal tissue engineering. Herein, a novel method to produce MTJ-like hydrogel yarns using a microfluidics-assisted 3D rotary wet-spinning strategy is developed. Optimization of flow rates, rotational speed, and delivery time of bioinks enables the production of highly compartmentalized scaffolds that recapitulate the muscle, tendon, and the transient MTJ-like region. Additionally, such biofabrication parameters are validated in terms of cellular response by promoting an optimal uniaxial alignment for both muscle and tendon precursor cells. By sequentially wet-spinning C2C12 myoblasts and NIH 3T3 fibroblasts, a gradient-patterned cellular arrangement mirroring the intrinsic biological heterogeneity of the MTJ is successfully obtained. The immunofluorescence assessment further reveals the localized expression of tissue-specific markers, including myosin heavy chain and collagen type I/III, which demonstrate muscle and tenogenic tissue maturation, respectively. Remarkably, the muscle-tendon transition zone exhibits finger-like projection of the multinucleated myotubes in the tenogenic compartment, epitomizing the MTJ signature architecture.

20.
Bioact Mater ; 42: 257-269, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39285913

ABSTRACT

The healing of large skin defects remains a significant challenge in clinical settings. The lack of epidermal sources, such as autologous skin grafting, limits full-thickness skin defect repair and leads to excessive scar formation. Skin organoids have the potential to generate a complete skin layer, supporting in-situ skin regeneration in the defect area. In this study, skin organoid spheres, created with human keratinocytes, fibroblasts, and endothelial cells, showed a specific structure with a stromal core surrounded by surface keratinocytes. We selected an appropriate bioink and innovatively combined an extrusion-based bioprinting technique with dual-photo source cross-linking technology to ensure the overall mechanical properties of the 3D bioprinted skin organoid. Moreover, the 3D bioprinted skin organoid was customized to match the size and shape of the wound site, facilitating convenient implantation. When applied to full-thickness skin defects in immunodeficient mice, the 3D bioprinted human-derived skin organoid significantly accelerated wound healing through in-situ regeneration, epithelialization, vascularization, and inhibition of excessive inflammation. The combination of skin organoid and 3D bioprinting technology can overcome the limitations of current skin substitutes, offering a novel treatment strategy to address large-area skin defects.

SELECTION OF CITATIONS
SEARCH DETAIL